Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetes ; 70(2): 423-435, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33154069

RESUMEN

The aim for this study was to elucidate how the hypothalamic hunger-inducing hormone acyl-ghrelin (AG), which is also produced in the pancreas, affects ß-cell function, with particular attention to the role of ATP-sensitive K+ (KATP) channels and the exact site of action of the hormone. AG hyperpolarized the membrane potential and decreased cytoplasmic calcium concentration [Ca2+]c and glucose-stimulated insulin secretion (GSIS). These effects were abolished in ß-cells from SUR1-knockout (KO) mice. AG increased KATP current but only in a configuration with intact metabolism. Unacylated ghrelin counteracted the effects of AG. The influence of AG on membrane potential and GSIS could only be averted in the combined presence of a ghrelin receptor (GHSR1a) antagonist and an inverse agonist. The inhibition of GSIS by AG could be prevented by dibutyryl cyclic-cAMP or 3-isobutyl-1-methylxanthine and the somatostatin (SST) receptor 2-5 antagonist H6056. These data indicate that AG indirectly opens KATP channels probably by interference with the cAMP/cAMP-dependent protein kinase pathway, resulting in a decrease of [Ca2+]c and GSIS. The experiments with SUR1-KO ß-cells point to a direct effect of AG on ß-cells and not, as earlier suggested, to an exclusive effect by AG-induced SST release from δ-cells. Nevertheless, SST receptors may be involved in the effect of AG, possibly by heteromerization of AG and SST receptors.


Asunto(s)
Ghrelina/análogos & derivados , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Canales KATP/metabolismo , Animales , Calcio/metabolismo , Ghrelina/farmacología , Glucosa/farmacología , Células Secretoras de Insulina/metabolismo , Ratones , Receptores de Somatostatina/metabolismo , Transducción de Señal/efectos de los fármacos , Somatostatina/metabolismo
2.
Front Endocrinol (Lausanne) ; 11: 545638, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193079

RESUMEN

Objective: Congenital hyperinsulinism (CHI) is a rare disease characterized by persistent hypoglycemia as a result of inappropriate insulin secretion, which can lead to irreversible neurological defects in infants. Poor efficacy and strong adverse effects of the current medications impede successful treatment. The aim of the study was to investigate new approaches to silence ß-cells and thus attenuate insulin secretion. Research Design and Methods: In the scope of our research, we tested substances more selective and more potent than the gold standard diazoxide that also interact with neuroendocrine ATP-sensitive K+ (KATP) channels. Additionally, KATP channel-independent targets as Ca2+-activated K+ channels of intermediate conductance (KCa3.1) and L-type Ca2+ channels were investigated. Experiments were performed using human islet cell clusters isolated from tissue of CHI patients (histologically classified as pathological) and islet cell clusters obtained from C57BL/6N (WT) or SUR1 knockout (SUR1-/-) mice. The cytosolic Ca2+ concentration ([Ca2+]c) was used as a parameter for the pathway regulated by electrical activity and was determined by fura-2 fluorescence. The mitochondrial membrane potential (ΔΨ) was determined by rhodamine 123 fluorescence and single channel currents were measured by the patch-clamp technique. Results: The selective KATP channel opener NN414 (5 µM) diminished [Ca2+]c in isolated human CHI islet cell clusters and WT mouse islet cell clusters stimulated with 10 mM glucose. In islet cell clusters lacking functional KATP channels (SUR1-/-) the drug was without effect. VU0071063 (30 µM), another KATP channel opener considered to be selective, lowered [Ca2+]c in human CHI islet cell clusters. The compound was also effective in islet cell clusters from SUR1-/- mice, showing that [Ca2+]c is influenced by additional effects besides KATP channels. Contrasting to NN414, the drug depolarized ΔΨ in murine islet cell clusters pointing to severe interference with mitochondrial metabolism. An opener of KCa3.1 channels, DCEBIO (100 µM), significantly decreased [Ca2+]c in SUR1-/- and human CHI islet cell clusters. To target L-type Ca2+ channels we tested two already approved drugs, dextromethorphan (DXM) and simvastatin. DXM (100 µM) efficiently diminished [Ca2+]c in stimulated human CHI islet cell clusters as well as in stimulated SUR1-/- islet cell clusters. Similar effects on [Ca2+]c were observed in experiments with simvastatin (7.2 µM). Conclusions: NN414 seems to provide a good alternative to the currently used KATP channel opener diazoxide. Targeting KCa3.1 channels by channel openers or L-type Ca2+ channels by DXM or simvastatin might be valuable approaches for treatment of CHI caused by mutations of KATP channels not sensitive to KATP channel openers.


Asunto(s)
Hiperinsulinismo Congénito/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Calcio/metabolismo , Bloqueadores de los Canales de Calcio/administración & dosificación , Células Cultivadas , Hiperinsulinismo Congénito/metabolismo , Óxidos S-Cíclicos/administración & dosificación , Dextrometorfano/administración & dosificación , Diazóxido , Humanos , Secreción de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Canales KATP/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Nifedipino/administración & dosificación
3.
Endocrine ; 68(3): 526-535, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32146655

RESUMEN

Novel agonists of the nuclear liver-X-receptor (LXR) are designed to treat metabolic disorders or cancer. The rationale to develop these new drugs is based on promising results with established LXR agonist like T0901317 and GW3965. LXRα and LXRß are expressed in ß-cells, and expression is increased by T0901317. The aim of the present study was to evaluate whether effects of these drugs on ß-cell function are specific and reliably linked to LXR activation. T0901317 and GW3965, widely used as specific LXR agonists, show rapid, non-genomic effects on stimulus-secretion coupling of mouse pancreatic ß-cells at low µM concentrations. T0901317 lowered the cytosolic Ca2+ concentration, reduced or completely inhibited action potentials, and decreased insulin secretion. GW3965 exerted similar effects on insulin secretion. T0901317 affected the production of reactive oxygen species and ATP. The involvement of the classical nuclear LXRs in T0901317- and GW3965-mediated effects in ß-cells could be ruled out using LXRα, LXRß and double knockout mice. Our results strongly suggest that LXR agonists, that are considered to be specific for this receptor, interfere with mitochondrial metabolism and metabolism-independent processes in ß-cells. Thus, it is indispensable to test novel LXR agonists accompanying to ongoing clinical trials for acute and chronic effects on cell function in cellular systems and/or animal models lacking classical LXRs.


Asunto(s)
Células Secretoras de Insulina , Receptores Nucleares Huérfanos , Animales , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Receptores X del Hígado , Ratones , Ratones Noqueados
4.
Endocrine ; 63(2): 270-283, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30229397

RESUMEN

PURPOSE: The role of ATP, which is secreted by pancreatic ß-cells, is still a matter of debate. It has been postulated that extracellular ATP acts as a positive auto- or paracrine signal in ß-cells amplifying insulin secretion. However, there is rising evidence that extracellular ATP may also mediate a negative signal. METHODS: We evaluated whether extracellular ATP interferes with the Ca2+-mediated negative feedback mechanism that regulates oscillatory activity of ß-cells. RESULTS: To experimentally uncover the Ca2+-induced feedback we applied a high extracellular Ca2+ concentration. Under this condition ATP (100 µM) inhibited glucose-evoked oscillations of electrical activity and hyperpolarized the membrane potential. Furthermore, ATP acutely increased the interburst phase of Ca2+ oscillations and reduced the current through L-type Ca2+ channels. Accordingly, ATP (500 µM) decreased glucose-induced insulin secretion. The ATP effect was not mimicked by AMP, ADP, or adenosine. The use of specific agonists and antagonists and mice deficient of large conductance Ca2+-dependent K+ channels revealed that P2X, but not P2Y receptors, and Ca2+-dependent K+ channels are involved in the underlying signaling cascade induced by ATP. The effectiveness of ATP to interfere with parameters of stimulus-secretion coupling is markedly reduced at low extracellular Ca2+ concentration. CONCLUSION: It is suggested that extracellular ATP which is co-secreted with insulin in a pulsatile manner during glucose-stimulated exocytosis provides a negative feedback signal driving ß-cell oscillations in co-operation with Ca2+ and other signals.


Asunto(s)
Adenosina Trifosfato/farmacología , Comunicación Autocrina/efectos de los fármacos , Glucosa/farmacología , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Animales , Calcio/metabolismo , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Insulina/farmacología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
Diabetes ; 68(2): 324-336, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30409782

RESUMEN

The Takeda-G-protein-receptor-5 (TGR5) mediates physiological actions of bile acids. Since it was shown that TGR5 is expressed in pancreatic tissue, a direct TGR5 activation in ß-cells is currently postulated and discussed. The current study reveals that oleanolic acid (OLA) affects murine ß-cell function by TGR5 activation. Both a Gαs inhibitor and an inhibitor of adenylyl cyclase (AC) prevented stimulating effects of OLA. Accordingly, OLA augmented the intracellular cAMP concentration. OLA and two well-established TGR5 agonists, RG239 and tauroursodeoxycholic acid (TUDCA), acutely promoted stimulus-secretion coupling (SSC). OLA reduced KATP current and elevated current through Ca2+ channels. Accordingly, in mouse and human ß-cells, TGR5 ligands increased the cytosolic Ca2+ concentration by stimulating Ca2+ influx. Higher OLA concentrations evoked a dual reaction, probably due to activation of a counterregulating pathway. Protein kinase A (PKA) was identified as a downstream target of TGR5 activation. In contrast, inhibition of phospholipase C and phosphoinositide 3-kinase did not prevent stimulating effects of OLA. Involvement of exchange protein directly activated by cAMP 2 (Epac2) or farnesoid X receptor (FXR2) was ruled out by experiments with knockout mice. The proposed pathway was not influenced by local glucagon-like peptide 1 (GLP-1) secretion from α-cells, shown by experiments with MIN6 cells, and a GLP-1 receptor antagonist. In summary, these data clearly demonstrate that activation of TGR5 in ß-cells stimulates insulin secretion via an AC/cAMP/PKA-dependent pathway, which is supposed to interfere with SSC by affecting KATP and Ca2+ currents and thus membrane potential.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Calcio/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Masculino , Ratones , Ácido Oleanólico/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Ácido Tauroquenodesoxicólico/farmacología
6.
J Biol Chem ; 294(10): 3707-3719, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30587573

RESUMEN

Neuroendocrine-type ATP-sensitive K+ (KATP) channels are metabolite sensors coupling membrane potential with metabolism, thereby linking insulin secretion to plasma glucose levels. They are octameric complexes, (SUR1/Kir6.2)4, comprising sulfonylurea receptor 1 (SUR1 or ABCC8) and a K+-selective inward rectifier (Kir6.2 or KCNJ11). Interactions between nucleotide-, agonist-, and antagonist-binding sites affect channel activity allosterically. Although it is hypothesized that opening these channels requires SUR1-mediated MgATP hydrolysis, we show here that ATP binding to SUR1, without hydrolysis, opens channels when nucleotide antagonism on Kir6.2 is minimized and SUR1 mutants with increased ATP affinities are used. We found that ATP binding is sufficient to switch SUR1 alone between inward- or outward-facing conformations with low or high dissociation constant, KD , values for the conformation-sensitive channel antagonist [3H]glibenclamide ([3H]GBM), indicating that ATP can act as a pure agonist. Assembly with Kir6.2 reduced SUR1's KD for [3H]GBM. This reduction required the Kir N terminus (KNtp), consistent with KNtp occupying a "transport cavity," thus positioning it to link ATP-induced SUR1 conformational changes to channel gating. Moreover, ATP/GBM site coupling was constrained in WT SUR1/WT Kir6.2 channels; ATP-bound channels had a lower KD for [3H]GBM than ATP-bound SUR1. This constraint was largely eliminated by the Q1179R neonatal diabetes-associated mutation in helix 15, suggesting that a "swapped" helix pair, 15 and 16, is part of a structural pathway connecting the ATP/GBM sites. Our results suggest that ATP binding to SUR1 biases KATP channels toward open states, consistent with SUR1 variants with lower KD values causing neonatal diabetes, whereas increased KD values cause congenital hyperinsulinism.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores de Sulfonilureas/química , Receptores de Sulfonilureas/metabolismo , Adenosina Difosfato/metabolismo , Regulación Alostérica , Animales , Sitios de Unión , Cricetinae , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Hidrólisis , Activación del Canal Iónico , Modelos Moleculares , Mutación , Canales de Potasio de Rectificación Interna/química , Unión Proteica , Conformación Proteica en Hélice alfa
7.
Pflugers Arch ; 470(3): 537-547, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29218453

RESUMEN

We have shown previously that genetic or pharmacological deletion of KATP channels protect against beta cell dysfunction induced by reactive oxygen species (ROS). Since it is assumed that glucolipotoxicity (GLTx) causes ROS production, we aimed to evaluate whether suppression of KATP channel activity can also prevent beta cell damage evoked by GLTx. We used an in vitro model of GLTx and measured distinct parameters of stimulus-secretion coupling. GLTx gradually induced disturbances of Ca2+ oscillations over 3 days. This impairment in Ca2+ dynamics was partially reversed in beta cells without functional KATP channels (SUR1-/-) and by the sulfonylurea gliclazide but not by tolbutamide. By contrast, the GLTx-induced suppression of glucose-induced insulin secretion could not be rescued by decreased KATP channel activity pointing to a direct interaction of GLTx with the secretory capacity. Accordingly, GLTx also suppressed KCl-induced insulin secretion. GLTx was not accompanied by decisively increased ROS production or enhanced apoptosis. Insulin content of beta cells was markedly reduced by GLTx, an effect not prevented by gliclazide. Since GLTx markedly diminished the mitochondrial membrane potential and cellular ATP content, lack of ATP is assumed to decrease insulin biosynthesis. The deleterious effect of GLTx is therefore caused by direct interference with the secretory capacity whereby reduction of insulin content is one important parameter. These findings deepen our understanding how GLTx damages beta cells and reveal that GLTx is disconnected from ROS formation, a notion important for targeting beta cells in the treatment of diabetes. Overall, GLTx-induced energy depletion may be a primary step in the cascade of events leading to loss of beta cell function in type-2 diabetes mellitus.


Asunto(s)
Apoptosis , Metabolismo Energético , Células Secretoras de Insulina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Gliclazida/farmacología , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Canales KATP/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Tolbutamida/farmacología
8.
Diabetes ; 66(11): 2840-2848, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28864549

RESUMEN

Atrial natriuretic peptide (ANP) influences glucose homeostasis and possibly acts as a link between the cardiovascular system and metabolism, especially in metabolic disorders like diabetes. The current study evaluated effects of ANP on ß-cell function by the use of a ß-cell-specific knockout of the ANP receptor with guanylate cyclase activity (ßGC-A-KO). ANP augmented insulin secretion at the threshold glucose concentration of 6 mmol/L and decreased KATP single-channel activity in ß-cells of control mice but not of ßGC-A-KO mice. In wild-type ß-cells but not ß-cells lacking functional KATP channels (SUR1-KO), ANP increased electrical activity, suggesting no involvement of other ion channels. At 6 mmol/L glucose, ANP readily elicited Ca2+ influx in control ß-cells. This effect was blunted in ß-cells of ßGC-A-KO mice, and the maximal cytosolic Ca2+ concentration was lower. Experiments with inhibitors of protein kinase G (PKG), protein kinase A (PKA), phosphodiesterase 3B (PDE3B), and a membrane-permeable cyclic guanosine monophosphate (cGMP) analog on KATP channel activity and insulin secretion point to participation of the cGMP/PKG and cAMP/PKA/Epac (exchange protein directly activated by cAMP) directly activated by cAMP Epac pathways in the effects of ANP on ß-cell function; the latter seems to prevail. Moreover, ANP potentiated the effect of glucagon-like peptide 1 (GLP-1) on glucose-induced insulin secretion, which could be caused by a cGMP-mediated inhibition of PDE3B, which in turn reduces cAMP degradation.


Asunto(s)
Factor Natriurético Atrial/farmacología , Glucosa/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/fisiología , Animales , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , GMP Cíclico/genética , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Tolbutamida
9.
Endocrinology ; 158(7): 2145-2154, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28449117

RESUMEN

The role of liver X receptor (LXR) in pancreatic ß-cell physiology and pathophysiology is still unclear. It has been postulated that chronic LXR activation in ß-cells induces lipotoxicity, a key step in the development of ß-cell dysfunction, which accompanies type 2 diabetes mellitus. In most of these studies, the LXR ligand T0901317 has been administered chronically in the micromolar range to study the significance of LXR activation. In the current study, we have evaluated acute effects of T0901317 on stimulus-secretion coupling of ß-cells. We found that 10 µM T0901317 completely suppressed oscillations of the cytosolic Ca2+ concentration induced by 15 mM glucose. Obviously, this effect was due to inhibition of mitochondrial metabolism. T0901317 markedly depolarized the mitochondrial membrane potential, thus inhibiting adenosine triphosphate (ATP) production and reducing the cytosolic ATP concentration. This led in turn to a huge increase in KATP current and hyperpolarization of the cell membrane potential. Eventually, T0901317 inhibited glucose-induced insulin secretion. These effects were rapid in on-set and not compatible with the activation of a nuclear receptor. In vivo, T0901317 acutely increased the blood glucose concentration after intraperitoneal application. In summary, these data clearly demonstrate that T0901317 exerts acute effects on stimulus-secretion coupling. This observation questions the chronic use of T0901317 and limits the interpretation of results obtained under these experimental conditions.


Asunto(s)
Hidrocarburos Fluorados/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Receptores X del Hígado/agonistas , Mitocondrias/efectos de los fármacos , Sulfonamidas/farmacología , Animales , Femenino , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/ultraestructura , Ligandos , Receptores X del Hígado/genética , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
10.
Endocrinology ; 157(12): 4677-4690, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27715254

RESUMEN

The angiotensin-converting enzyme 2/angiotensin (Ang)-(1-7)/Mas axis of the renin-angiotensin system often opposes the detrimental effects of the angiotensin-converting enzyme/Ang II/Ang II type 1 receptor axis and has been associated with beneficial effects on glucose homeostasis, whereas underlying mechanisms are mostly unknown. Here we investigate the effects of Ang-(1-7) and its receptor Mas on ß-cell function. Isolated islets from Mas-deficient and wild-type mice were stimulated with Ang-(1-7) or its antagonists and effects on insulin secretion determined. Islets' cytoplasmic calcium and cAMP concentrations, mRNA amounts of Ins1, Ins2, Pdx1, and Mafa and effects of inhibitors of cAMP downstream signaling were determined. Ang-(1-7) was also applied to mice by osmotic pumps for 14 days and effects on glucose tolerance and insulin secretion were assessed. Ang-(1-7) increased insulin secretion from wild-type islets, whereas antagonists and genetic Mas deficiency led to reduced insulin secretion. The Mas-dependent effects of Ang-(1-7) on insulin secretion did not result from changes in insulin gene expression or changes in the excitation-secretion coupling but from increased intracellular cAMP involving exchange protein activated directly by cAMP. Administration of Ang-(1-7) in vivo had only marginal effects on glucose tolerance in wild-type mice but still resulted in improved insulin secretion from islets isolated of these mice. Interestingly, although less pronounced than in wild types, Ang-(1-7) still affected insulin secretion in Mas-deficient islets. The data indicate a significant function of Ang-(1-7) in the regulation of insulin secretion from mouse islets in vitro and in vivo, mainly, but not exclusively, by Mas-dependent signaling, modulating the accessory pathway of insulin secretion via increase in cAMP.


Asunto(s)
Angiotensina I/farmacología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , AMP Cíclico/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Insulina/genética , Resistencia a la Insulina/fisiología , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Factores de Transcripción Maf de Gran Tamaño/genética , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones , Ratones Noqueados , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Transactivadores/genética , Transactivadores/metabolismo
11.
Pflugers Arch ; 467(11): 2389-97, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25893711

RESUMEN

Pancreatic beta-cells respond to an unchanging stimulatory glucose concentration with oscillations in membrane potential (Vm), cytosolic Ca(2+) concentration ([Ca(2+)]c), and insulin secretion. The underlying mechanisms are largely ascertained. Some particular details, however, are still in debate. Stimulus-secretion coupling (SSC) of beta-cells comprises glucose-induced Ca(2+) influx into the cytosol and thus into mitochondria. It is suggested that this activates (mitochondrial) dehydrogenases leading to an increase in reduction equivalents and ATP production. According to SSC, a glucose-induced increase in ATP production would thus further augment ATP production, i.e. induce a feed-forward loop that is hardly compatible with oscillations. Consistently, other studies favour a feedback mechanism that drives oscillatory mitochondrial ATP production. If Ca(2+) influx activates dehydrogenases, a change in [Ca(2+)]c should increase the concentration of reduction equivalents. We measured changes in flavin adenine dinucleotide (FAD) and nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) autofluorescence in response to changes in glucose concentration or glucose-independent changes in [Ca(2+)]c. The FAD signal was altered by glucose but not by alterations in [Ca(2+)]c. NAD(P)H was increased by glucose but even decreased by Ca(2+) influx evoked by tolbutamide. The mitochondrial membrane potential ΔΨ was hyperpolarized by 4 mM glucose. As adding tolbutamide then depolarized ΔΨ, we deduce that Ca(2+) does not activate mitochondrial activity but by contrast even inhibits it by reducing the driving force for ATP production. Inhibition of Ca(2+) influx reversed the Ca(2+)-induced changes in ΔΨ and NAD(P)H. The results are consistent with a feedback mechanism which transiently and repeatedly reduces ATP production and explain the oscillatory activity of pancreatic beta-cells at increased glucose concentrations.


Asunto(s)
Calcio/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Oxidorreductasas/metabolismo , Animales , FMN Reductasa/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Glucosa/farmacología , Hipoglucemiantes/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tolbutamida/farmacología
12.
Diabetologia ; 58(7): 1532-41, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25874444

RESUMEN

AIMS/HYPOTHESIS: Generation of reduction equivalents is a prerequisite for nutrient-stimulated insulin secretion. Mitochondrial succinate dehydrogenase (SDH) fulfils a dual function with respect to mitochondrial energy supply: (1) the enzyme is part of mitochondrial respiratory chains; and (2) it catalyses oxidation of succinate to fumarate in the Krebs cycle. The aim of our study was to elucidate the significance of SDH for beta cell stimulus-secretion coupling (SSC). METHODS: Mitochondrial variables, reactive oxygen species (ROS) and cytosolic Ca(2+) concentration ([Ca(2+)]c) were measured by fluorescence techniques and insulin release by radioimmunoassay in islets or islet cells of C57Bl/6N mice. RESULTS: Inhibition of SDH with 3-nitropropionic acid (3-NPA) or monoethyl fumarate (MEF) reduced glucose-stimulated insulin secretion. Inhibition of the ATP-sensitive K(+) channel (KATP channel) partly prevented this effect, whereas potentiation of antioxidant defence by superoxide dismutase mimetics (TEMPOL and mito-TEMPO) or by nuclear factor erythroid 2-related factor 2 (Nrf-2)-mediated upregulation of antioxidant enzymes (oltipraz, tert-butylhydroxyquinone) did not diminish the inhibitory influence of 3-NPA. Blocking SDH decreased glucose-stimulated increase in intracellular FADH2 concentration without alterations in NAD(P)H. In addition, 3-NPA and MEF drastically reduced glucose-induced hyperpolarisation of mitochondrial membrane potential, indicative of decreased ATP production. As a consequence, the glucose-stimulated rise in [Ca(2+)]c was significantly delayed and reduced. Acute application of 3-NPA interrupted glucose-driven oscillations of [Ca(2+)]c. 3-NPA per se did not elevate intracellular ROS, but instead prevented glucose-induced ROS accumulation. CONCLUSIONS/INTERPRETATION: SDH is an important regulator of insulin secretion and ROS production. Inhibition of SDH interrupts membrane-potential-dependent SSC, pointing to a pivotal role of mitochondrial FAD/FADH2 homeostasis for the maintenance of glycaemic control.


Asunto(s)
Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Mitocondrias/enzimología , Especies Reactivas de Oxígeno/metabolismo , Succinato Deshidrogenasa/metabolismo , Animales , Calcio/metabolismo , Inhibidores Enzimáticos/farmacología , Flavina-Adenina Dinucleótido/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Canales KATP/efectos de los fármacos , Canales KATP/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Nitrocompuestos/farmacología , Propionatos/farmacología , Succinato Deshidrogenasa/antagonistas & inhibidores , Compuestos de Sulfonilurea/farmacología
13.
Endocrinology ; 156(4): 1263-71, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25599407

RESUMEN

We have recently shown that the bile acid (BA) taurochenodeoxycholate (TCDC) acutely stimulates insulin secretion via activation of the farnesoid X receptor (FXR). Aims of the current investigation were to discriminate between nongenomic (≤1 h) and genomic effects (24-48 h) of BAs on ß-cells and to evaluate whether FXR can modulate the adverse effects of a high-fat diet (HFD). TCDC (500 nM) as well as glycine-conjugated and unconjugated CDC (chenodeoxycholate) increased insulin secretion in acute incubations but did not evoke additional effects after 1-2 days of preincubation. The BAs did not stimulate ß-cells of FXR-knockout (KO) mice and activation of the G protein-coupled BA receptor TGR5 was ineffective, suggesting that FXR is the sole BA receptor in ß-cells activated by TCDC and its analogues. As opposed to lean mice, obese FXR-KO mice did not show HFD-induced glucose intolerance and increased fasting glucose. The beneficial impact of FXR-KO on glucose metabolism cannot be explained by an adaptive compensation of insulin secretion or ß-cell mass. Interestingly, in contrast to its effect on islets from lean mice, the FXR agonist GW4064 was ineffective in stimulating insulin secretion of islets from wild type mice fed a HFD or isolated islets kept in a glucolipotoxic medium. Additional feeding of CDC restored the effect of GW4064. CDC prevented HFD-induced impairment of glucose tolerance and in vitro effects of glucolipotoxicity. The data show that the FXR is the most important BA receptor in ß-cells and that FXR signaling in ß-cells is impaired by overnutrition, which alters activatability of the FXR.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácido Quenodesoxicólico/farmacología , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Isoxazoles/farmacología , Masculino , Ratones , Ratones Noqueados , Ratones Obesos , Obesidad/genética , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética , Ácido Tauroquenodesoxicólico/farmacología
14.
Integr Biol (Camb) ; 6(5): 540-4, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24658612

RESUMEN

Extracellular recording of the glucose-induced electrical activity of mouse islets of Langerhans on microelectrode arrays (MEAs) is an innovative and powerful tool to address beta-cell (patho-)physiology. In a dual approach we tested whether this technique can detect concentration-dependent drug effects as well as characterize alterations in beta-cell activity during prolonged culture. First we established conditions that allow long-term investigation of beta-cell function by recording electrical activity. The results provide the first measurements of beta-cell membrane potential oscillations of individual murine islets during long-term culture. Oscillations were recorded for up to 34 days after islet isolation. Importantly, the glucose dependence of electrical activity did not change over a period of one month. Thus we can follow electrophysiological changes of individual islets induced by alterations in the beta-cell environment over weeks. Second, we used the MEA technique to assay beta-cell damage induced by oxidative stress and to evaluate appropriate protection mechanisms. Oxidative stress plays a key role in the development of type 2 diabetes mellitus (T2DM). Examination of the acute effects of H2O2 on electrical activity showed that the oxidant reduced the electrical activity in a concentration-dependent manner. The superoxide dismutase mimetic, tempol, protected against the detrimental effects of H2O2. In conclusion, we demonstrated that MEA recordings can be used to address disease-related mechanisms and protective interventions in beta-cells. In the future, this fundamental work should enable the monitoring of the electrical activity of islets of Langerhans under controlled ex vivo conditions including long-term exposure to oxidative stress, glucolipotoxicity, and other diabetes-inducing agents.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Islotes Pancreáticos/metabolismo , Potenciales de la Membrana/fisiología , Estrés Oxidativo/fisiología , Animales , Antioxidantes/farmacología , Óxidos N-Cíclicos/farmacología , Relación Dosis-Respuesta a Droga , Electrofisiología/métodos , Peróxido de Hidrógeno/antagonistas & inhibidores , Peróxido de Hidrógeno/metabolismo , Ratones Endogámicos C57BL , Microelectrodos , Marcadores de Spin
15.
Mol Pharmacol ; 83(1): 51-60, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23024191

RESUMEN

Earlier studies suggest that glitazones exert beneficial effects in patients with type 2 diabetes by directly affecting insulin secretion of ß-cells, besides improving the effectiveness of insulin in peripheral tissues. The effects of glitazones on stimulus-secretion coupling (SSC) are poorly understood. We tested the influence of troglitazone and pioglitazone on different parameters of SSC, including insulin secretion (radioimmunoassay), cell membrane potential, various ion currents (patch-clamp), mitochondrial membrane potential (ΔΨ), and cytosolic Ca(2+) concentration (fluorescence). Troglitazone exerted stimulatory, inhibitory, or no effects on insulin secretion depending on the drug and glucose concentration. It depolarized the ΔΨ, thus lowering ATP production, which resulted in opening of ATP-dependent K(+) channels (K(ATP) channels) and reduced insulin secretion. However, it also exerted direct inhibitory effects on K(ATP) channels that can explain enhanced insulin secretion. Troglitazone also inhibited the currents through voltage-dependent Ca(2+) and K(+) channels. Pioglitazone was less effective than troglitazone on all parameters tested. The effects of both glitazones were markedly reduced in the presence of bovine serum albumin. Glitazones exert multiple actions on ß-cell SSC that have to be considered as undesired side effects because the influence of these compounds on ß-cells is not controllable. The final effect on insulin secretion depends on many parameters, including the actual glucose and drug concentration, protein binding of the drug, and the drug by itself. Troglitazone and pioglitazone differ in their influence on SSC. It can be assumed that the effects of pioglitazone on ß-cells are negligible under in vivo conditions.


Asunto(s)
Cromanos/farmacología , Hipoglucemiantes/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Tiazolidinedionas/farmacología , Animales , Calcio/metabolismo , Canales de Calcio Tipo L/fisiología , Bovinos , Células Cultivadas , Citosol/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Canales KATP/fisiología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Pioglitazona , Canales de Potasio con Entrada de Voltaje/fisiología , Radioinmunoensayo , Albúmina Sérica Bovina/farmacología , Troglitazona
16.
Islets ; 4(5): 333-8, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23073079

RESUMEN

Bile acids (BAs) are important signaling molecules that are involved in the regulation of their own metabolism, lipid metabolism, energy expenditure and glucose homeostasis. The nuclear farnesoid X receptor (FXR) and the G-protein-coupled TGR-5 are the most prominent BA receptors. FXR is highly expressed in liver and activation of liver FXR profoundly affects glucose homeostasis. Strikingly, the effect of FXR activation on glucose metabolism seems to depend on the nutritional status of the organism, i.e., slimness or obesity. Recently, it became evident that FXR is present in pancreatic ß cells and that activation of ß cell FXR contributes to the regulation of glucose homeostasis. Interestingly, FXR activation increases glucose-induced insulin secretion by non-genomic effects on stimulus-secretion coupling. The first chapter of this review shortly introduces the role of liver FXR in glucose metabolism, the second part focuses on the impact of FXR in lean and obese animals, and the third chapter highlights the significance of FXR in ß cells.


Asunto(s)
Células Secretoras de Insulina/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Glucemia/análisis , Humanos , Obesidad/metabolismo
17.
Free Radic Biol Med ; 53(8): 1574-83, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22922339

RESUMEN

The cGMP-dependent protein kinase I (cGKI) is a key mediator of cGMP signaling, but the specific functions of its two isoforms, cGKIα and cGKIß, are poorly understood. Recent studies indicated a novel cGMP-independent role for cGKIα in redox sensing. To dissect the effects of oxidative stress on the cGKI isoforms, we used mouse embryonic fibroblasts and vascular smooth muscle cells (VSMCs) expressing both, one, or none of them. In cGKIα-expressing cells, but not in cells expressing only cGKIß, incubation with H2O2 induced the formation of a disulfide bond between the two identical subunits of the dimeric enzyme. Oxidation of cGKIα was associated with increased phosphorylation of its substrate, vasodilator-stimulated phosphoprotein. H2O2 did not stimulate cGMP production, indicating that it activates cGKIα directly via oxidation. Interestingly, there was a mutual influence of H2O2 and cGMP on cGKI activity and disulfide bond formation, respectively; preoxidation of the kinase with H2O2 slightly impaired its activation by cGMP, whereas preactivation of the enzyme with cGMP attenuated its oxidation by H2O2. To evaluate the functional relevance of the noncanonical H2O2-cGKIα pathway, we studied the regulation of the cytosolic Ca²âº concentration ([Ca²âº](i)). H2O2 suppressed norepinephrine-induced Ca²âº transients in cGKIα-expressing VSMCs and, to a lower extent, in VSMCs expressing only cGKIß or none of the isoforms. Thus, H2O2 lowers [Ca²âº](i) mainly via a cGKIα-dependent pathway. These results indicate that oxidative stress selectively targets the cGKIα isoform, which then modulates cellular processes in a cGMP-independent manner. A decrease in [Ca²âº](i) in VSMCs via activation of cGKIα might be a major mechanism of H2O2-induced vasodilation.


Asunto(s)
Calcio/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Citosol/metabolismo , Embrión de Mamíferos/enzimología , Fibroblastos/enzimología , Peróxido de Hidrógeno/farmacología , Músculo Liso Vascular/enzimología , Animales , Western Blotting , Células Cultivadas , GMP Cíclico/metabolismo , Disulfuros/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Oxidantes/farmacología , Transducción de Señal
18.
Diabetes ; 61(6): 1479-89, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22492528

RESUMEN

Type 2 diabetes mellitus is associated with alterations in bile acid (BA) signaling. The aim of our study was to test whether pancreatic ß-cells contribute to BA-dependent regulation of glucose homeostasis. Experiments were performed with islets from wild-type, farnesoid X receptor (FXR) knockout (KO), and ß-cell ATP-dependent K(+) (K(ATP)) channel gene SUR1 (ABCC8) KO mice, respectively. Sodium taurochenodeoxycholate (TCDC) increased glucose-induced insulin secretion. This effect was mimicked by the FXR agonist GW4064 and suppressed by the FXR antagonist guggulsterone. TCDC and GW4064 stimulated the electrical activity of ß-cells and enhanced cytosolic Ca(2+) concentration ([Ca(2+)](c)). These effects were blunted by guggulsterone. Sodium ursodeoxycholate, which has a much lower affinity to FXR than TCDC, had no effect on [Ca(2+)](c) and insulin secretion. FXR activation by TCDC is suggested to inhibit K(ATP) current. The decline in K(ATP) channel activity by TCDC was only observed in ß-cells with intact metabolism and was reversed by guggulsterone. TCDC did not alter insulin secretion in islets of SUR1-KO or FXR-KO mice. TCDC did not change islet cell apoptosis. This is the first study showing an acute action of BA on ß-cell function. The effect is mediated by FXR by nongenomic elements, suggesting a novel link between FXR activation and K(ATP) channel inhibition.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Ácido Taurodesoxicólico/farmacología , Animales , Glucosa/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Noqueados , Canales de Potasio de Rectificación Interna/genética , Pregnenodionas/farmacología , Receptores Citoplasmáticos y Nucleares/genética
19.
Pflugers Arch ; 462(6): 835-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21947556

RESUMEN

The membrane potential (V (m)) of beta-cells oscillates at glucose concentrations between ~6 and 25 mM, i.e. burst phases with action potentials alternate with silent interburst phases generating so-called slow waves. The slow waves drive oscillations of the cytosolic Ca(2+) concentration ([Ca(2+)](c)) and insulin secretion. The length of the bursts correlates with the amount of insulin release. Thus, the fraction of plateau phase (FOPP), i.e. the percentage of time with burst activity, is an excellent marker for beta-cell function and metabolic integrity. Extracellular voltage changes of mouse islets were measured using a microelectrode array (MEA) allowing the detection of burst and interburst phases. At a non-stimulating glucose concentration (3 mM) no electrical activity was detectable while bursting was continuous at 30 mM. The glucose concentration-response (determined as FOPP) curve revealed half-maximal stimulation at 12 ± 1 mM (Hill equation fit). The signal was sensitive to K(ATP) channel modulators, e.g. tolbutamide or diazoxide. Simultaneous recordings of electrical activity and [Ca(2+)](c) revealed congruent bursts and peaks, respectively. The extracellular recordings are in perfect agreement with more time-consuming intracellular electrical recordings. The results provide a 'proof-of-principle' for detection of beta-cell slow waves and determination of the FOPP using extracellular electrodes in a MEA-based system. The method is facile and provides the capability to study the effects of modulators of beta-cell function including possible anti-diabetic drugs in real time. Moreover, the method may be useful for checking the metabolic integrity of human donor islets prior to transplantation.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Potenciales de la Membrana/fisiología , Microelectrodos , Técnicas de Placa-Clamp/instrumentación , Técnicas de Placa-Clamp/métodos , Potenciales de Acción/fisiología , Animales , Antihipertensivos/farmacología , Calcio/metabolismo , Diazóxido , Glucosa/metabolismo , Humanos , Hipoglucemiantes/farmacología , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Tolbutamida/farmacología
20.
Islets ; 2(3): 156-63, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21099309

RESUMEN

The AMP-activated protein kinase (AMPK) is one of the key players in cellular energy regulation adapting cellular demands to nutritional and metabolic variations. Oral antidiabetic drugs like metformin and glitazones (thiazolidinediones) are known to stimulate this enzyme. Besides their established action on peripheral organs including liver and muscles, it has been claimed that these drugs may affect ß-cell function. However, it is still a matter of debate whether pharmacological AMPK stimulation increases or decreases insulin secretion. To study this point and to reveal mechanisms underlying changes in insulin secretion we used the specific AMPK activator AICAR and investigated its effects on stimulus-secretion coupling. Membrane potential and currents were measured by the patch-clamp technique, [Ca (2+)]c, mitochondrial membrane potential, and NAD(P)H by fluorescence techniques and insulin secretion by a radioimmunoassay. AICAR enhanced glucose-stimulated insulin release, an effect that can be attributed to the augmentation of electrical activity and [Ca (2+)]c resulting from an AICAR-evoked inhibition of the KATP current. This latter effect was not due to a direct interaction of AICAR with the K[ATP] channels but was dependent on cell metabolism. AICAR did not affect mitochondrial membrane potential or NAD(P)H autofluorescence. Metformin mimicked the action of AICAR on electrical activity, [Ca (2+) ]c, and K[ATP] current. However, compared to AICAR the effects were less pronounced and not sufficient to stimulate insulin secretion. In conclusion, activation of AMPK augments nutrient-induced insulin secretion. Thus, targeting AMPK of ß-cells may be an appropriate strategy for the treatment of disturbed glucose homeostasis..


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Activación Enzimática/fisiología , Hipoglucemiantes/farmacología , Secreción de Insulina , Células Secretoras de Insulina/enzimología , Células Secretoras de Insulina/metabolismo , Canales KATP/efectos de los fármacos , Canales KATP/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metformina/farmacología , Ratones , Ratones Endogámicos C57BL , NADP/metabolismo , Técnicas de Placa-Clamp , Ribonucleótidos/farmacología , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...